1
|
Bülbül B, Ding K, Zhan CG, Çiftçi G, Yelekçi K, Gürboğa M, Özakpınar ÖB, Aydemir E, Baybağ D, Şahin F, Kulabaş N, Helvacıoğlu S, Charehsaz M, Tatar E, Özbey S, Küçükgüzel İ. Novel 1,2,4-triazoles derived from Ibuprofen: synthesis and in vitro evaluation of their mPGES-1 inhibitory and antiproliferative activity. Mol Divers 2023; 27:2185-2215. [PMID: 36331786 DOI: 10.1007/s11030-022-10551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Some novel triazole-bearing ketone and oxime derivatives were synthesized from Ibuprofen. In vitro cytotoxic activities of all synthesized molecules against five cancer lines (human breast cancer MCF-7, human lung cancer A549, human prostate cancer PC-3, human cervix cancer HeLa, and human chronic myelogenous leukemia K562 cell lines) were evaluated by MTT assay. In addition, mouse embryonic fibroblast cells (NIH/3T3) were also evaluated to determine the selectivity. Compounds 18, 36, and 45 were found to be the most cytotoxic, and their IC50 values were in the range of 17.46-68.76 µM, against the tested cancer cells. According to the results, compounds 7 and 13 demonstrated good anti-inflammatory activity against the microsomal enzyme prostaglandin E2 synthase-1 (mPGES-1) enzyme at IC50 values of 13.6 and 4.95 µM. The low cytotoxicity and non-mutagenity of these compounds were found interesting. Also, these compounds significantly prevented tube formation in angiogenesis studies. In conclusion, the anti-inflammatory and angiogenesis inhibitory activities of these compounds without toxicity suggested that they may be promising agents in anti-inflammatory treatment and they may be supportive agents for the cancer treatment.
Collapse
Affiliation(s)
- Bahadır Bülbül
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Düzce University, Konuralp, Düzce, Turkey
- Department of Pharmaceutical Chemistry, Institute of Health Sciences, Marmara University, Dragos, Kartal, 34865, Istanbul, Turkey
| | - Kai Ding
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Gamze Çiftçi
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey
| | - Kemal Yelekçi
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey
| | - Merve Gürboğa
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, Istanbul, Turkey
| | - Özlem Bingöl Özakpınar
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, Istanbul, Turkey
| | - Esra Aydemir
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayışdağı, Istanbul, Turkey
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Biruni University, Zeytinburnu, 34010, Turkey
| | - Deniz Baybağ
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayışdağı, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayışdağı, Istanbul, Turkey
| | - Necla Kulabaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, 34854, Istanbul, Turkey
| | - Sinem Helvacıoğlu
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Ataşehir, 34750, Istanbul, Turkey
| | - Mohammad Charehsaz
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Ataşehir, 34750, Istanbul, Turkey
| | - Esra Tatar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, 34854, Istanbul, Turkey
| | - Süheyla Özbey
- Department of Physics Engineering, Faculty of Engineering, Hacettepe University, Beytepe, 06800, Ankara, Turkey
| | - İlkay Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, 34854, Istanbul, Turkey.
| |
Collapse
|
2
|
Synthesis, in vitro and in silico studies on novel 3-aryloxymethyl-5-[(2-oxo-2-arylethyl)sulfanyl]-1,2,4-triazoles and their oxime derivatives as potent inhibitors of mPGES-1. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
3
|
Finetti F, Travelli C, Ercoli J, Colombo G, Buoso E, Trabalzini L. Prostaglandin E2 and Cancer: Insight into Tumor Progression and Immunity. BIOLOGY 2020; 9:E434. [PMID: 33271839 PMCID: PMC7760298 DOI: 10.3390/biology9120434] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022]
Abstract
The involvement of inflammation in cancer progression has been the subject of research for many years. Inflammatory milieu and immune response are associated with cancer progression and recurrence. In different types of tumors, growth and metastatic phenotype characterized by the epithelial mesenchymal transition (EMT) process, stemness, and angiogenesis, are increasingly associated with intrinsic or extrinsic inflammation. Among the inflammatory mediators, prostaglandin E2 (PGE2) supports epithelial tumor aggressiveness by several mechanisms, including growth promotion, escape from apoptosis, transactivation of tyrosine kinase growth factor receptors, and induction of angiogenesis. Moreover, PGE2 is an important player in the tumor microenvironment, where it suppresses antitumor immunity and regulates tumor immune evasion, leading to increased tumoral progression. In this review, we describe the current knowledge on the pro-tumoral activity of PGE2 focusing on its role in cancer progression and in the regulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Federica Finetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, 27100 Pavia, Italy; (C.T.); (E.B.)
| | - Jasmine Ercoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Erica Buoso
- Department of Pharmaceutical Sciences, University of Pavia, 27100 Pavia, Italy; (C.T.); (E.B.)
| | - Lorenza Trabalzini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
4
|
Kirkby NS, Raouf J, Ahmetaj-Shala B, Liu B, Mazi SI, Edin ML, Chambers MG, Korotkova M, Wang X, Wahli W, Zeldin DC, Nüsing R, Zhou Y, Jakobsson PJ, Mitchell JA. Mechanistic definition of the cardiovascular mPGES-1/COX-2/ADMA axis. Cardiovasc Res 2020; 116:1972-1980. [PMID: 31688905 PMCID: PMC7519887 DOI: 10.1093/cvr/cvz290] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/23/2019] [Accepted: 10/31/2019] [Indexed: 02/05/2023] Open
Abstract
AIMS Cardiovascular side effects caused by non-steroidal anti-inflammatory drugs (NSAIDs), which all inhibit cyclooxygenase (COX)-2, have prevented development of new drugs that target prostaglandins to treat inflammation and cancer. Microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors have efficacy in the NSAID arena but their cardiovascular safety is not known. Our previous work identified asymmetric dimethylarginine (ADMA), an inhibitor of endothelial nitric oxide synthase, as a potential biomarker of cardiovascular toxicity associated with blockade of COX-2. Here, we have used pharmacological tools and genetically modified mice to delineate mPGES-1 and COX-2 in the regulation of ADMA. METHODS AND RESULTS Inhibition of COX-2 but not mPGES-1 deletion resulted in increased plasma ADMA levels. mPGES-1 deletion but not COX-2 inhibition resulted in increased plasma prostacyclin levels. These differences were explained by distinct compartmentalization of COX-2 and mPGES-1 in the kidney. Data from prostanoid synthase/receptor knockout mice showed that the COX-2/ADMA axis is controlled by prostacyclin receptors (IP and PPARβ/δ) and the inhibitory PGE2 receptor EP4, but not other PGE2 receptors. CONCLUSION These data demonstrate that inhibition of mPGES-1 spares the renal COX-2/ADMA pathway and define mechanistically how COX-2 regulates ADMA.
Collapse
Affiliation(s)
- Nicholas S Kirkby
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| | - Joan Raouf
- Unit of Rheumatology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Blerina Ahmetaj-Shala
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| | - Bin Liu
- Cardiovascular Research Centre, Shantou University Medical College, Shantou, China
| | - Sarah I Mazi
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
- King Fahad Cardiac Center, King Saud University, Riyadh, Saudi Arabia
| | - Matthew L Edin
- National Institute for Environmental Health Sciences, Durham, NC, USA
| | | | - Marina Korotkova
- Unit of Rheumatology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xiaomeng Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology & Research, Singapore, Singapore
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, UK
- Singapore Eye Research Institute, Singapore, Singapore
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Darryl C Zeldin
- National Institute for Environmental Health Sciences, Durham, NC, USA
| | - Rolf Nüsing
- Clinical Pharmacology and Pharmacotherapy Department, Goethe University, Frankfurt, Germany
| | - Yingbi Zhou
- Cardiovascular Research Centre, Shantou University Medical College, Shantou, China
| | - Per-Johan Jakobsson
- Unit of Rheumatology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Jane A Mitchell
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
5
|
Woolbright BL, Pilbeam CC, Taylor JA. Prostaglandin E2 as a therapeutic target in bladder cancer: From basic science to clinical trials. Prostaglandins Other Lipid Mediat 2020; 148:106409. [PMID: 31931078 DOI: 10.1016/j.prostaglandins.2020.106409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/02/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022]
Abstract
Bladder cancer (BCa) is a common solid tumor marked by high rates of recurrence, especially in non-muscle invasive disease. Prostaglandin E2 (PGE2) is a ubiquitously present lipid mediator responsible for numerous physiological actions. Inhibition of cyclooxygenase (COX) enzymes by the non-steroidal anti-inflammatory (NSAID) class of drugs results in reduced PGE2 levels. NSAID usage has been associated with reductions in cancers such as BCa. Clinical trials using NSAIDs to prevent recurrence have had mixed results, but largely converge on issues with cardiotoxicity. The purpose of this review is to understand the basic science behind how and why inhibitors of PGE2 may be effective against BCa, and to explore alternate therapeutic modalities for addressing the role of PGE2 without the associated cardiotoxicity. We will address the role of PGE2 in a diverse array of cancer-related functions including stemness, immunosuppression, proliferation, cellular signaling and more.
Collapse
Affiliation(s)
| | - Carol C Pilbeam
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
6
|
Zhang Q, Li S, Ye G, Wang M, Ni L, Kan S, Zhang M, Chen J. Prostaglandin E2 receptor EP2 mediates the effect of cyclooxygenase 2 on secondary parathyroid hyperplasia in end-stage renal disease. Nephrol Dial Transplant 2020; 34:606-617. [PMID: 29982796 DOI: 10.1093/ndt/gfy194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Secondary hyperparathyroidism (SHPT) in patients with end-stage renal disease (ESRD) is characterized by hyperplasia of the parathyroid glands (PTGs), while the underlying mechanism is not completely understood. Previously we demonstrated a relationship between cyclooxygenase 2 (COX2) overexpression and parathyroid hyperplasia and here we investigate the role of COX2 downstream metabolic product prostaglandin E2 (PGE2) and its receptor EP2 in the pathogenesis of SHPT. METHODS PTGs isolated from ESRD patients with advanced SHPT were used to test the expression of COX2-microsomal prostaglandin E synthase-1 (mPGES-1)-EP2 pathway. A diffuse proliferative section of the PTGs was used for tissue culture and treated with high phosphate (HPi) medium, COX2-PGE2-EP2 pathway inhibitors or agonists. EP2 short hairpin RNA (shRNA) lentivirus was locally applied to treat an SHPT rat model. RESULTS In PTGs isolated from ESRD patients, enhanced immunoactivities of COX2, mPGES-1 and EP2 were observed. In primary cultured PTG tissues, HPi induced intact parathyroid hormone (iPTH) secretion, proliferating cell nuclear antigen (PCNA) expression and COX2 activity, while COX2 and EP2 inhibitors attenuated hyperparathyroidism promoted by HPi. Furthermore, PGE2 or EP2 agonist (butaprost) directly stimulated hyperparathyroidism, whereas EP2 receptor antagonist or cyclic adenosine monophosphate inhibitor attenuated the hyperparathyroidism promoted by PGE2 or butaprost. EP2 shRNA treatment significantly reduced excessive expressions of EP2 and PCNA in the PTGs of nephrectomy rats fed an HPi diet, diminished the size of PTGs and downregulated serum iPTH levels. CONCLUSIONS The COX2 downstream PGE2 and its receptor EP2 may play an important role in HPi-induced parathyroid hyperplasia and may serve as a potential therapeutic target for SHPT in ESRD.
Collapse
Affiliation(s)
- Qian Zhang
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shensen Li
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoxin Ye
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengjing Wang
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Ni
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sen Kan
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minmin Zhang
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Chen
- Division of Nephrology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Larsson K, Steinmetz J, Bergqvist F, Arefin S, Spahiu L, Wannberg J, Pawelzik SC, Morgenstern R, Stenberg P, Kublickiene K, Korotkova M, Jakobsson PJ. Biological characterization of new inhibitors of microsomal PGE synthase-1 in preclinical models of inflammation and vascular tone. Br J Pharmacol 2019; 176:4625-4638. [PMID: 31404942 DOI: 10.1111/bph.14827] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/18/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Microsomal PGE synthase-1 (mPGES-1), the inducible synthase that catalyses the terminal step in PGE2 biosynthesis, is of high interest as therapeutic target to treat inflammation. Inhibition of mPGES-1 is suggested to be safer than traditional NSAIDs, and recent data demonstrate anti-constrictive effects on vascular tone, indicating new therapeutic opportunities. However, there is a lack of potent mPGES-1 inhibitors lacking interspecies differences for conducting in vivo studies in relevant preclinical disease models. EXPERIMENTAL APPROACH Potency was determined based on the reduction of PGE2 formation in recombinant enzyme assays, cellular assay, human whole blood assay, and air pouch mouse model. Anti-inflammatory properties were assessed by acute paw swelling in a paw oedema rat model. Effect on vascular tone was determined with human ex vivo wire myography. KEY RESULTS We report five new mPGES-1 inhibitors (named 934, 117, 118, 322, and 323) that selectively inhibit recombinant human and rat mPGES-1 with IC50 values of 10-29 and 67-250 nM respectively. The compounds inhibited PGE2 production in a cellular assay (IC50 values 0.15-0.82 μM) and in a human whole blood assay (IC50 values 3.3-8.7 μM). Moreover, the compounds blocked PGE2 formation in an air pouch mouse model and reduced acute paw swelling in a paw oedema rat model. Human ex vivo wire myography analysis showed reduced adrenergic vasoconstriction after incubation with the compounds. CONCLUSION AND IMPLICATIONS These mPGES-1 inhibitors can be used as refined tools in further investigations of the role of mPGES-1 in inflammation and microvascular disease.
Collapse
Affiliation(s)
- Karin Larsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Julia Steinmetz
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Filip Bergqvist
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Linda Spahiu
- Biochemical Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Wannberg
- SciLifeLab Drug Discovery and Development Platform, Medicinal Chemistry-Lead Identification, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sven-Christian Pawelzik
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Theme Heart and Vessels, Division of Valvular and Coronary Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Ralf Morgenstern
- Biochemical Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Marina Korotkova
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
A review on mPGES-1 inhibitors: From preclinical studies to clinical applications. Prostaglandins Other Lipid Mediat 2019; 147:106383. [PMID: 31698145 DOI: 10.1016/j.prostaglandins.2019.106383] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
Prostaglandin E2 (PGE2) is a lipid mediator of inflammation and cancer progression. It is mainly formed via metabolism of arachidonic acid by cyclooxygenases (COX) and the terminal enzyme microsomal prostaglandin E synthase-1 (mPGES-1). Widely used non-steroidal anti-inflammatory drugs (NSAIDs) inhibit COX activity, resulting in decreased PGE2 production and symptomatic relief. However, NSAIDs block the production of many other lipid mediators that have important physiological and resolving actions, and these drugs cause gastrointestinal bleeding and/or increase the risk for severe cardiovascular events. Selective inhibition of downstream mPGES-1 for reduction in only PGE2 biosynthesis is suggested as a safer therapeutic strategy. This review covers the recent advances in characterization of new mPGES-1 inhibitors in preclinical models and their future clinical applications.
Collapse
|
9
|
Montrose DC, Galluzzi L. Drugging cancer metabolism: Expectations vs. reality. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 347:1-26. [PMID: 31451211 DOI: 10.1016/bs.ircmb.2019.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As compared to their normal counterparts, neoplastic cells exhibit a variety of metabolic changes that reflect not only genetic and epigenetic defects underlying malignant transformation, but also the nutritional and immunobiological conditions of the tumor microenvironment. Such alterations, including the so-called Warburg effect (an increase in glucose uptake largely feeding anabolic and antioxidant metabolism), have attracted considerable attention as potential targets for the development of novel anticancer therapeutics. However, very few drugs specifically conceived to target bioenergetic cancer metabolism are currently approved by regulatory agencies for use in humans. This reflects the elevated degree of heterogeneity and redundancy in the metabolic circuitries exploited by neoplastic cells from different tumors (even of the same type), as well as the resemblance of such metabolic pathways to those employed by highly proliferating normal cells. Here, we summarize the major metabolic alterations that accompany oncogenesis, the potential of targeting bioenergetic metabolism for cancer therapy, and the obstacles that still prevent the clinical translation of such a promising therapeutic paradigm.
Collapse
Affiliation(s)
- David C Montrose
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université Paris Descartes/Paris V, Paris, France.
| |
Collapse
|
10
|
Bergqvist F, Ossipova E, Idborg H, Raouf J, Checa A, Englund K, Englund P, Khoonsari PE, Kultima K, Wheelock CE, Larsson K, Korotkova M, Jakobsson PJ. Inhibition of mPGES-1 or COX-2 Results in Different Proteomic and Lipidomic Profiles in A549 Lung Cancer Cells. Front Pharmacol 2019; 10:636. [PMID: 31231223 PMCID: PMC6567928 DOI: 10.3389/fphar.2019.00636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/17/2019] [Indexed: 12/23/2022] Open
Abstract
Pharmacological inhibition of microsomal prostaglandin E synthase (mPGES)-1 for selective reduction in prostaglandin E2 (PGE2) biosynthesis is protective in experimental models of cancer and inflammation. Targeting mPGES-1 is envisioned as a safer alternative to traditional non-steroidal anti-inflammatory drugs (NSAIDs). Herein, we compared the effects of mPGES-1 inhibitor Compound III (CIII) with the cyclooxygenase (COX)-2 inhibitor NS-398 on protein and lipid profiles in interleukin (IL)-1β-induced A549 lung cancer cells using mass spectrometry. Inhibition of mPGES-1 decreased PGE2 production and increased PGF2α and thromboxane B2 (TXB2) formation, while inhibition of COX-2 decreased the production of all three prostanoids. Our proteomics results revealed that CIII downregulated multiple canonical pathways including eIF2, eIF4/P70S6K, and mTOR signaling, compared to NS-398 that activated these pathways. Moreover, pathway analysis predicted that CIII increased cell death of cancer cells (Z = 3.8, p = 5.1E-41) while NS-398 decreased the same function (Z = -5.0, p = 6.5E-35). In our lipidomics analyses, we found alterations in nine phospholipids between the two inhibitors, with a stronger alteration in the lysophospholipid (LPC) profile with NS-398 compared to CIII. Inhibition of mPGES-1 increased the concentration of sphinganine and dihydroceramide (C16:0DhCer), while inhibition of COX-2 caused a general decrease in most ceramides, again suggesting different effects on cell death between the two inhibitors. We showed that CIII decreased proliferation and potentiated the cytotoxic effect of the cytostatic drugs cisplatin, etoposide, and vincristine when investigated in a live cell imaging system. Our results demonstrate differences in protein and lipid profiles after inhibition of mPGES-1 or COX-2 with important implications on the therapeutic potential of mPGES-1 inhibitors as adjuvant treatment in cancer. We encourage further investigations to illuminate the clinical benefit of mPGES-1 inhibitors in cancer.
Collapse
Affiliation(s)
- Filip Bergqvist
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Elena Ossipova
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Helena Idborg
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Joan Raouf
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Antonio Checa
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karin Englund
- Department of Analytical Chemistry, Stockholm University, Stockholm, Sweden
| | - Petter Englund
- Department of Analytical Chemistry, Stockholm University, Stockholm, Sweden
| | - Payam Emami Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Marina Korotkova
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
11
|
Wang T, Jing B, Sun B, Liao Y, Song H, Xu D, Guo W, Li K, Hu M, Liu S, Ling J, Kuang Y, Feng Y, Zhou BP, Deng J. Stabilization of PTGES by deubiquitinase USP9X promotes metastatic features of lung cancer via PGE 2 signaling. Am J Cancer Res 2019; 9:1145-1160. [PMID: 31285948 PMCID: PMC6610053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/09/2019] [Indexed: 06/09/2023] Open
Abstract
Early metastasis and local recurrence are the major causes of mortality and poor prognosis of non-small cell lung cancer (NSCLC). However, the underlying mechanisms of these processes are poorly understood. In this study, we aimed to investigate the roles of the PTGES/PGE2 pathway in lung cancer progression. We found that prostaglandin E synthase (PTGES), a key enzyme for PGE2 synthesis in the arachidonic acid pathway, was highly dysregulated in NSCLC. Dysregulated PTGES was essential for the promotion of tumor migration and metastasis of NSCLC cells. Knockdown of PTGES in lung cancer cells resulted in suppressed cell migration, which was reversed by exogenous PGE2. Consistent with this, PTGES knockdown also reduced the expression of CSC markers, tumor sphere formation, colony forming activity, tumorigenicity, and lung metastasis in vivo. Dysregulated PTGES is mainly attributed to protein stabilization by USP9X, a deubiquitination enzyme. USP9X physically interacted with PTGES and prevented it from proteasome-directed degradation via deubiquitination. Consistent with this, USP9X expression was highly correlated with PTGES expression in NSCLC tumor tissues. Taken together, our results show that the upregulated USP9X-PTGES-PGE2 axis contributes significantly to the metastatic features of NSCLC.
Collapse
Affiliation(s)
- Tong Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Bo Jing
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Beibei Sun
- Translational Medical Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Yueling Liao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Hongyong Song
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Dongliang Xu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Wenzheng Guo
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Kaimi Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Min Hu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Shuli Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, The Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Jing Ling
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yanbin Kuang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Yao Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky College of MedicineLexington, KY, USA
| | - Jiong Deng
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of MedicineShanghai, China
- Translational Medical Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
12
|
Zhang X, Feng Y, Liu X, Ma J, Li Y, Wang T, Li X. Beyond a chemopreventive reagent, aspirin is a master regulator of the hallmarks of cancer. J Cancer Res Clin Oncol 2019; 145:1387-1403. [PMID: 31037399 DOI: 10.1007/s00432-019-02902-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/22/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Aspirin, one of the most commonly used nonsteroidal anti-inflammatory drugs (NAIDS), not only shows cancer chemoprevention effects but also improves cancer therapeutic effects when combined with other therapies. Studies that focus on aspirin regulation of the hallmarks of cancer and the associated molecular mechanisms facilitate a more thorough understanding of aspirin in mediating chemoprevention and may supply additional information for the development of novel cancer therapeutic agents. METHODS The relevant literatures from PubMed have been reviewed in this article. RESULTS Current studies have revealed that aspirin regulates almost all the hallmarks of cancer. Within tumor tissue, aspirin suppresses the bioactivities of cancer cells themselves and deteriorates the tumor microenvironment that supports cancer progression. In addition to tumor tissues, blocking of platelet activation also contributes to the ability of aspirin to inhibit cancer progression. In terms of the molecular mechanism, aspirin targets oncogenes and cancer-related signaling pathways and activates certain tumor suppressors. CONCLUSION Beyond a chemopreventive agent, aspirin is a master regulator of the hallmarks of cancer.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150086, China
| | - Yukuan Feng
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xi Liu
- Center of Cardiovascular Disease, Inner Mongolia People's Hospital, Hohhot, 010017, Inner Mongolia, China
| | - Jianhui Ma
- Department of Pathology, Harbin Medical University, Harbin, 150086, China
| | - Yafei Li
- Department of Pathology, Harbin Medical University, Harbin, 150086, China
| | - Tianzhen Wang
- Department of Pathology, Harbin Medical University, Harbin, 150086, China.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
13
|
HARA S. Prostaglandin terminal synthases as novel therapeutic targets. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:703-723. [PMID: 29129850 PMCID: PMC5743848 DOI: 10.2183/pjab.93.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 07/21/2017] [Indexed: 06/07/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) exert their anti-inflammatory and anti-tumor effects by reducing prostaglandin (PG) production via the inhibition of cyclooxygenase (COX). However, the gastrointestinal, renal and cardiovascular side effects associated with the pharmacological inhibition of the COX enzymes have focused renewed attention onto other potential targets for NSAIDs. PGH2, a COX metabolite, is converted to each PG species by species-specific PG terminal synthases. Because of their potential for more selective modulation of PG production, PG terminal synthases are now being investigated as a novel target for NSAIDs. In this review, I summarize the current understanding of PG terminal synthases, with a focus on microsomal PGE synthase-1 (mPGES-1) and PGI synthase (PGIS). mPGES-1 and PGIS cooperatively exacerbate inflammatory reactions but have opposing effects on carcinogenesis. mPGES-1 and PGIS are expected to be attractive alternatives to COX as therapeutic targets for several diseases, including inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Shuntaro HARA
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Tokyo, Japan
| |
Collapse
|
14
|
Olesch C, Sha W, Angioni C, Sha LK, Açaf E, Patrignani P, Jakobsson PJ, Radeke HH, Grösch S, Geisslinger G, von Knethen A, Weigert A, Brüne B. MPGES-1-derived PGE2 suppresses CD80 expression on tumor-associated phagocytes to inhibit anti-tumor immune responses in breast cancer. Oncotarget 2016; 6:10284-96. [PMID: 25871398 PMCID: PMC4496355 DOI: 10.18632/oncotarget.3581] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 02/13/2015] [Indexed: 01/04/2023] Open
Abstract
Prostaglandin E2 (PGE2) favors multiple aspects of tumor development and immune evasion. Therefore, microsomal prostaglandin E synthase (mPGES-1/-2), is a potential target for cancer therapy. We explored whether inhibiting mPGES-1 in human and mouse models of breast cancer affects tumor-associated immunity. A new model of breast tumor spheroid killing by human PBMCs was developed. In this model, tumor killing required CD80 expression by tumor-associated phagocytes to trigger cytotoxic T cell activation. Pharmacological mPGES-1 inhibition increased CD80 expression, whereas addition of PGE2, a prostaglandin E2 receptor 2 (EP2) agonist, or activation of signaling downstream of EP2 reduced CD80 expression. Genetic ablation of mPGES-1 resulted in markedly reduced tumor growth in PyMT mice. Macrophages of mPGES-1−/− PyMT mice indeed expressed elevated levels of CD80 compared to their wildtype counterparts. CD80 expression in tumor-spheroid infiltrating mPGES-1−/− macrophages translated into antigen-specific cytotoxic T cell activation. In conclusion, mPGES-1 inhibition elevates CD80 expression by tumor-associated phagocytes to restrict tumor growth. We propose that mPGES-1 inhibition in combination with immune cell activation might be part of a therapeutic strategy to overcome the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Catherine Olesch
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Weixiao Sha
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Carlo Angioni
- Institute of Clinical Pharmacology/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Lisa Katharina Sha
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Elias Açaf
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences and Center of Excellence on Aging (CeSI), "G. d'Annunzio" University, Chieti, Italy
| | - Per-Johan Jakobsson
- Department of Medicine, Rheumatology Research Unit, Karolinska Institutet, Stockholm, Sweden
| | - Heinfried H Radeke
- Pharmazentrum Frankfurt/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Sabine Grösch
- Institute of Clinical Pharmacology/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
15
|
Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-diclofenac as an anti-cancer agent. Ecancermedicalscience 2016; 10:610. [PMID: 26823679 PMCID: PMC4720497 DOI: 10.3332/ecancer.2016.610] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Diclofenac (DCF) is a well-known and widely used non-steroidal anti-inflammatory drug (NSAID), with a range of actions which are of interest in an oncological context. While there has long been an interest in the use of NSAIDs in chemoprevention, there is now emerging evidence that such drugs may have activity in a treatment setting. DCF, which is a potent inhibitor of COX-2 and prostaglandin E2 synthesis, displays a range of effects on the immune system, the angiogenic cascade, chemo- and radio-sensitivity and tumour metabolism. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. Based on this evidence the case is made for further clinical investigation of the anticancer effects of DCF, particularly in combination with other agents - with a range of possible multi-drug and multi-modality combinations outlined in the supplementary materials accompanying the main paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | | | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
16
|
Guo B, Tian XC, Li DD, Yang ZQ, Cao H, Zhang QL, Liu JX, Yue ZP. Expression, regulation and function of Egr1 during implantation and decidualization in mice. Cell Cycle 2015; 13:2626-40. [PMID: 25486203 DOI: 10.4161/15384101.2014.943581] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract Early growth response gene 1 (Egr1), a zinc finger transcriptional factor, plays an important role in regulating cell proliferation, differentiation and angiogenesis. Current data have shown that Egr1 is involved in follicular development, ovulation, luteinization and placental angiogenesis. However, the expression, regulation and function of Egr1 in mouse uterus during embryo implantation and decidualization are poorly understood. Here we showed that Egr1 was strongly expressed in the subluminal stroma surrounding the implanting blastocyst on day 5 of pregnancy. Injection of Egr1 siRNA into the mouse uterine horn could obviously reduce the number of implanted embryos and affect the uterine vascular permeability. Further study found that Egr1 played a role through influencing the expression of cyclooxygenase-2 (Cox-2), microsomal prostaglandin E synthase 1 (mPGES-1), vascular endothelial growth factor (Vegf), transformation related protein 53 (Trp53) and matrix metallopeptidase 9 (Mmp9) genes in the process of mouse embryo implantation. Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) might direct the expression of Egr1 in the uterine stromal cells. Under in vivo and in vitro artificial decidualization, Egr1 expression was significantly decreased. Overexpression of Egr1 downregulated the expression of decidual marker decidual/trophoblast PRL-related protein (Dtprp) in the uterine stromal cells, while inhibition of Egr1 upregulated the expression of Dtprp under in vitro decidualization. Estrogen and progesterone could regulate the expression of Egr1 in the ovariectomized mouse uterus and uterine stromal cells. These results suggest that Egr1 may be essential for embryo implantation and decidualization.
Collapse
Affiliation(s)
- Bin Guo
- a College of Veterinary Medicine ; Jilin University ; Changchun , P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Larsson K, Jakobsson PJ. Inhibition of microsomal prostaglandin E synthase-1 as targeted therapy in cancer treatment. Prostaglandins Other Lipid Mediat 2015; 120:161-5. [PMID: 26100239 DOI: 10.1016/j.prostaglandins.2015.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/22/2015] [Accepted: 06/02/2015] [Indexed: 11/29/2022]
|
18
|
Luz JG, Antonysamy S, Kuklish SL, Condon B, Lee MR, Allison D, Yu XP, Chandrasekhar S, Backer R, Zhang A, Russell M, Chang SS, Harvey A, Sloan AV, Fisher MJ. Crystal Structures of mPGES-1 Inhibitor Complexes Form a Basis for the Rational Design of Potent Analgesic and Anti-Inflammatory Therapeutics. J Med Chem 2015; 58:4727-37. [DOI: 10.1021/acs.jmedchem.5b00330] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- John Gately Luz
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Stephen Antonysamy
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Steven L. Kuklish
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Bradley Condon
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Matthew R. Lee
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Dagart Allison
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Xiao-Peng Yu
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Srinivasan Chandrasekhar
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Ryan Backer
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Aiping Zhang
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Marijane Russell
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Shawn S. Chang
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, Suite 200, San Diego, California 92121, United States
| | - Anita Harvey
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Ashley V. Sloan
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Matthew J. Fisher
- Lilly Research
Laboratories, Lilly Corporate Center, 355 East Merrill Street, Indianapolis, Indiana 46285, United States
| |
Collapse
|
19
|
Angulo J, Cuevas P, Cuevas B, El Youssef M, Fernández A, Martínez-Salamanca E, González-Corrochano R, Giménez-Gallego G. Diacetyloxyl derivatization of the fibroblast growth factor inhibitor dobesilate enhances its anti-inflammatory, anti-angiogenic and anti-tumoral activities. J Transl Med 2015; 13:48. [PMID: 25638171 PMCID: PMC4318172 DOI: 10.1186/s12967-015-0413-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/20/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Dobesilate (2,5-dihydroxyphenyl sulfonate, DHPS) was recently identified as the most potent member of a family of fibroblast growth factor (FGF) inhibitors headed by gentisic acid, one of the main catabolites of aspirin. Although FGFs were first described as inducers of angiogenesis, they were soon recognized as broad spectrum mitogens. Furthermore, in the last decade these proteins have been shown to participate directly in the onset of inflammation, and their potential angiogenic activity often contributes to the inflammatory process in vivo. The aim of this work was to evaluate the anti-inflammatory, anti-angiogenic and anti-tumoral activities of the derivative of DHPS obtained by acetoxylation of its two hydroxyl groups (2,5-diacetoxyphenyl sulfonate; DAPS). METHODS Anti-inflammatory, anti-angiogenic and anti-tumoral activities of DHPS and DAPS were compared using in vivo assays of dermatitis, angiogenesis and tumorigenesis. The effects of both compounds on myeloperoxidase (MPO) and cyclooxygenase (COX) activities, cytokine production and FGF-induced fibroblast proliferation were also determined. RESULTS Topical DAPS is more effective than DHPS in preventing inflammatory signs (increased vascular permeability, edema, leukocyte infiltration, MPO activation) caused by contact dermatitis induction in rat ears. DAPS, but not DHPS, effectively inhibits COX-1 and COX-2 activities. DAPS also reduces the increase in serum cytokine concentration induced by lipopolysaccharide in rats. Furthermore, DAPS displays higher in vivo efficacy than DHPS in inhibiting FGF-induced angiogenesis and heterotopic glioma progression, with demonstrated oral efficacy to combat both processes. CONCLUSIONS By inhibiting both FGF-signaling and COX-mediated prostaglandin synthesis, DAPS efficiently breaks the vicious circle created by the reciprocal induction of FGF and prostaglandins, which probably sustains undesirable inflammation in many circumstances. Our findings define the enhancement of anti-inflammatory, anti-angiogenic and anti-tumoral activities by diacetyloxyl derivatization of the FGF inhibitor, dobesilate.
Collapse
Affiliation(s)
- Javier Angulo
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - Pedro Cuevas
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - Begoña Cuevas
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - Mohammad El Youssef
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - Argentina Fernández
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - Eduardo Martínez-Salamanca
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - Rocío González-Corrochano
- Departamento de Estructura y Función de Proteínas, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Avda Ramiro de Maeztu 9, ES-28040, Madrid, Spain.
| | - Guillermo Giménez-Gallego
- Departamento de Estructura y Función de Proteínas, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Avda Ramiro de Maeztu 9, ES-28040, Madrid, Spain.
| |
Collapse
|
20
|
Nakanishi M, Perret C, Meuillet EJ, Rosenberg DW. Non-cell autonomous effects of targeting inducible PGE2 synthesis during inflammation-associated colon carcinogenesis. Carcinogenesis 2015; 36:478-86. [PMID: 25634334 DOI: 10.1093/carcin/bgv004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/14/2015] [Indexed: 12/21/2022] Open
Abstract
Microsomal PGE2 synthase-1 (mPGES-1), the terminal enzyme in the formation of inducible PGE2, represents a potential target for cancer chemoprevention. We have previously shown that genetic abrogation of mPGES-1 significantly suppresses tumorigenesis in two preclinical models of intestinal cancer. In this study, we examined the role of mPGES-1 during colon tumorigenesis in the presence of dextran sulfate sodium (DSS)-induced inflammatory microenvironment. Using Apc (Δ14/+) in which the mPGES-1 gene is either wild-type (D14:WT) or deleted (D14:KO), we report that mPGES-1 deficiency enhances sensitivity to acute mucosal injury. As a result of the increased epithelial damage, protection against adenoma formation is unexpectedly compromised in the D14:KO mice. Examining the DSS-induced acute injury, cryptal structures are formed within inflamed areas of colonic mucosa of both genotypes that display the hallmarks of early neoplasia. When acute epithelial injury is balanced by titration of DSS exposures, however, these small cryptal lesions progress rapidly to adenomas in the D14:WT mice. Given that mPGES-1 is highly expressed within the intestinal stroma under the inflammatory conditions of DSS-induced ulceration, we propose a complex and dual role for inducible PGE2 synthesis within the colonic mucosa. Our data suggest that inducible PGE2 is critical for the maintenance of an intact colonic epithelial barrier, while promoting epithelial regeneration. This function is exploited during neoplastic transformation in Apc (Δ14/+) mice as PGE2 contributes to the growth and expansion of the early initiated cryptal structures. Taken together, inducible PGE2 plays a complex role in inflammation-associated cancers that requires further analysis. Inducible PGE2 production by mPGES-1 is critical for the colonic mucosal homeostasis. This function is exploited in the presence of the neoplastic transformation in Apc (Δ14/+) mice as PGE2 contributes to the growth and expansion of the early cryptal structures.
Collapse
Affiliation(s)
- Masako Nakanishi
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA, Inserm, U1016, département endocrinologie métabolisme et cancer, Institut Cochin, Paris, France and The University of Arizona Cancer Center, Department of Molecular and Cell Biology, Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA. Tucson, AZ, USA
| | - Christine Perret
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA, Inserm, U1016, département endocrinologie métabolisme et cancer, Institut Cochin, Paris, France and
| | - Emmanuelle J Meuillet
- The University of Arizona Cancer Center, Department of Molecular and Cell Biology, Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA. Tucson, AZ, USA
| | - Daniel W Rosenberg
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA, Inserm, U1016, département endocrinologie métabolisme et cancer, Institut Cochin, Paris, France and The University of Arizona Cancer Center, Department of Molecular and Cell Biology, Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA. Tucson, AZ, USA
| |
Collapse
|
21
|
Kekatpure VD, Singh M, Selvam S, Shetkar G, Hedne NC, Trivedi NP, Siddappa G, Govindan SV, Suresh A, Rangarajan B, Dannenberg AJ, Kuriakose MA. Factors predicting outcome after salvage treatment for stage IV oral squamous cell carcinoma: Evidence of the potential importance of the cyclooxygenase-2-prostaglandin E2 pathway. Head Neck 2014; 37:1142-9. [PMID: 24771596 DOI: 10.1002/hed.23721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 04/22/2014] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND We determined the clinicopathological factors that predicted outcome after salvage treatment for stage IV oral squamous cell carcinoma (OSCC). Additionally, the prognostic significance of the cyclooxygenase-2 (COX-2)/microsomal prostaglandin-E synthase-1 (mPGES-1) pathway was evaluated. METHODS Thirty-one patients who underwent salvage surgery were included. COX-2 and mPGES-1 levels were quantified by real time polymerase chain reaction (PCR). RESULTS The 2-year disease-free and overall survival rates were 46% and 53%, respectively. Adequacy of initial treatment, tobacco smoking, and the presence of pathological risk factors were predictive of mortality. In patients who had not received chemotherapy before salvage surgery, high levels of intratumoral COX-2 and mPGES-1 were associated with poor prognosis. By contrast, high intratumoral COX-2 and mPGES-1 after chemotherapy were associated with improved outcomes. CONCLUSION Clinicopathological factors may inform treatment decisions in patients with stage IV OSCC. Expression patterns of COX-2 and mPGES-1 correlated with outcome and warrant further investigation. © 2014 Wiley Periodicals, Inc. Head Neck 37: 1142-1149, 2015.
Collapse
Affiliation(s)
- Vikram D Kekatpure
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Mandeep Singh
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Sumithra Selvam
- Department of Biostatistics, St. John's Research Institute, Bangalore, India
| | - Girish Shetkar
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Naveen C Hedne
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Nirav P Trivedi
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Gangotri Siddappa
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Sindhu V Govindan
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Amritha Suresh
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Bharath Rangarajan
- Department of Medical Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| | - Andrew J Dannenberg
- Department of Medicine, Weill Medical College of Cornell University, New York, New York
| | - Moni Abraham Kuriakose
- Department of Head and Neck Oncology, Mazumdar-Shaw Cancer Center, Narayana Hrudayalaya, Health City, Bangalore, India
| |
Collapse
|
22
|
Jiang J, Dingledine R. Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection. Trends Pharmacol Sci 2013; 34:413-23. [PMID: 23796953 DOI: 10.1016/j.tips.2013.05.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/05/2013] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
Abstract
Modulation of a specific prostanoid synthase or receptor provides therapeutic alternatives to nonsteroidal anti-inflammatory drugs (NSAIDs) for treating pathological conditions governed by cyclooxygenase-2 (COX-2 or PTGS2). Among the COX-2 downstream signaling pathways, the prostaglandin E2 (PGE2) receptor EP2 subtype (PTGER2) is emerging as a crucial mediator of many physiological and pathological events. Genetic ablation strategies and recent advances in chemical biology provide tools for a better understanding of EP2 signaling. In the brain, the EP2 receptor modulates some beneficial effects, including neuroprotection, in acute models of excitotoxicity, neuroplasticity, and spatial learning via cAMP-PKA signaling. Conversely, EP2 activation accentuates chronic inflammation mainly through the cAMP-Epac pathway, likely contributing to delayed neurotoxicity. EP2 receptor activation also engages β-arrestin in a G-protein-independent pathway that promotes tumor cell growth and migration. Understanding the conditions under which multiple EP2 signaling pathways are engaged might suggest novel therapeutic strategies to target this key inflammatory prostaglandin receptor.
Collapse
Affiliation(s)
- Jianxiong Jiang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | |
Collapse
|